Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.558
Filtrar
1.
Sci Rep ; 14(1): 13782, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877073

RESUMEN

Kaposi's sarcoma (KS) is a cancer affecting skin and internal organs for which the Kaposi's sarcoma associated herpesvirus (KSHV) is a necessary cause. Previous work has pursued KS diagnosis by quantifying KSHV DNA in skin biopsies using a point-of-care (POC) device which performs quantitative loop-mediated isothermal amplification (LAMP). These previous studies revealed that extracting DNA from patient biopsies was the rate limiting step in an otherwise rapid process. In this study, a simplified, POC-compatible alkaline DNA extraction, ColdSHOT, was optimized for 0.75 mm human skin punch biopsies. The optimized ColdSHOT extraction consistently produced 40,000+ copies of DNA per 5 µl reaction from 3 mg samples-a yield comparable to standard spin column extractions-within 1 h without significant equipment. The DNA yield was estimated sufficient for KSHV detection from KS-positive patient biopsies, and the LAMP assay was not affected by non-target tissue in the unpurified samples. Furthermore, the yields achieved via ColdSHOT were robust to sample storage in phosphate-buffered saline (PBS) or Tris-EDTA (TE) buffer prior to DNA extraction, and the DNA sample was stable after extraction. The results presented in this study indicate that the ColdSHOT DNA extraction could be implemented to simplify and accelerate the LAMP-based diagnosis of Kaposi's sarcoma using submillimeter biopsy samples.


Asunto(s)
ADN Viral , Herpesvirus Humano 8 , Técnicas de Amplificación de Ácido Nucleico , Sarcoma de Kaposi , Piel , Humanos , ADN Viral/genética , ADN Viral/aislamiento & purificación , Herpesvirus Humano 8/aislamiento & purificación , Herpesvirus Humano 8/genética , Biopsia/métodos , Piel/virología , Piel/patología , Sarcoma de Kaposi/diagnóstico , Sarcoma de Kaposi/virología , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/genética , Técnicas de Amplificación de Ácido Nucleico/métodos , Sistemas de Atención de Punto , Técnicas de Diagnóstico Molecular/métodos
2.
Nat Commun ; 15(1): 4841, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844783

RESUMEN

Kaposi sarcoma associated herpesvirus (KSHV) is associated with around 1% of all human tumors, including the B cell malignancy primary effusion lymphoma (PEL), in which co-infection with the Epstein Barr virus (EBV) can almost always be found in malignant cells. Here, we demonstrate that KSHV/EBV co-infection of mice with reconstituted human immune systems (humanized mice) leads to IgM responses against both latent and lytic KSHV antigens, and expansion of central and effector memory CD4+ and CD8+ T cells. Among these, KSHV/EBV dual-infection allows for the priming of CD8+ T cells that are specific for the lytic KSHV antigen K6 and able to kill KSHV/EBV infected B cells. This suggests that K6 may represent a vaccine antigen for the control of KSHV and its associated pathologies in high seroprevalence regions, such as Sub-Saharan Africa.


Asunto(s)
Linfocitos B , Linfocitos T CD8-positivos , Herpesvirus Humano 8 , Animales , Herpesvirus Humano 8/inmunología , Humanos , Linfocitos B/inmunología , Ratones , Linfocitos T CD8-positivos/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Coinfección/inmunología , Coinfección/virología , Linfocitos T CD4-Positivos/inmunología , Herpesvirus Humano 4/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Inmunoglobulina M/inmunología , Antígenos Virales/inmunología , Ratones SCID , Linfoma de Efusión Primaria/inmunología , Linfoma de Efusión Primaria/virología , Anticuerpos Antivirales/inmunología
3.
J Med Virol ; 96(5): e29684, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38773828

RESUMEN

Kaposi's sarcoma (KS) may derive from Kaposi's sarcoma herpesvirus (KSHV)-infected human mesenchymal stem cells (hMSCs) that migrate to sites characterized by inflammation and angiogenesis, promoting the initiation of KS. By analyzing the RNA sequences of KSHV-infected primary hMSCs, we have identified specific cell subpopulations, mechanisms, and conditions involved in the initial stages of KSHV-induced transformation and reprogramming of hMSCs into KS progenitor cells. Under proangiogenic environmental conditions, KSHV can reprogram hMSCs to exhibit gene expression profiles more similar to KS tumors, activating cell cycle progression, cytokine signaling pathways, endothelial differentiation, and upregulating KSHV oncogenes indicating the involvement of KSHV infection in inducing the mesenchymal-to-endothelial (MEndT) transition of hMSCs. This finding underscores the significance of this condition in facilitating KSHV-induced proliferation and reprogramming of hMSCs towards MEndT and closer to KS gene expression profiles, providing further evidence of these cell subpopulations as precursors of KS cells that thrive in a proangiogenic environment.


Asunto(s)
Herpesvirus Humano 8 , Células Madre Mesenquimatosas , Sarcoma de Kaposi , Humanos , Herpesvirus Humano 8/fisiología , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/virología , Células Madre Mesenquimatosas/virología , Diferenciación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Proliferación Celular
4.
J Virol ; 98(6): e0000524, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38717113

RESUMEN

TRIM32 is often aberrantly expressed in many types of cancers. Kaposi's sarcoma-associated herpesvirus (KSHV) is linked with several human malignancies, including Kaposi's sarcoma and primary effusion lymphomas (PELs). Increasing evidence has demonstrated the crucial role of KSHV lytic replication in viral tumorigenesis. However, the role of TRIM32 in herpesvirus lytic replication remains unclear. Here, we reveal that the expression of TRIM32 is upregulated by KSHV in latency, and reactivation of KSHV lytic replication leads to the inhibition of TRIM32 in PEL cells. Strikingly, RTA, the master regulator of lytic replication, interacts with TRIM32 and dramatically promotes TRIM32 for degradation via the proteasome systems. Inhibition of TRIM32 induces cell apoptosis and in turn inhibits the proliferation and colony formation of KSHV-infected PEL cells and facilitates the reactivation of KSHV lytic replication and virion production. Thus, our data imply that the degradation of TRIM32 is vital for the lytic activation of KSHV and is a potential therapeutic target for KSHV-associated cancers. IMPORTANCE: TRIM32 is associated with many cancers and viral infections; however, the role of TRIM32 in viral oncogenesis remains largely unknown. In this study, we found that the expression of TRIM32 is elevated by Kaposi's sarcoma-associated herpesvirus (KSHV) in latency, and RTA (the master regulator of lytic replication) induces TRIM32 for proteasome degradation upon viral lytic reactivation. This finding provides a potential therapeutic target for KSHV-associated cancers.


Asunto(s)
Herpesvirus Humano 8 , Proteínas Inmediatas-Precoces , Transactivadores , Factores de Transcripción , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Replicación Viral , Humanos , Herpesvirus Humano 8/fisiología , Herpesvirus Humano 8/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Transactivadores/metabolismo , Transactivadores/genética , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Inmediatas-Precoces/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteolisis , Latencia del Virus , Apoptosis , Activación Viral , Sarcoma de Kaposi/virología , Sarcoma de Kaposi/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Línea Celular , Linfoma de Efusión Primaria/virología , Linfoma de Efusión Primaria/metabolismo
5.
Viruses ; 16(5)2024 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-38793630

RESUMEN

During viral infection, the innate immune system utilizes a variety of specific intracellular sensors to detect virus-derived nucleic acids and activate a series of cellular signaling cascades that produce type I IFNs and proinflammatory cytokines and chemokines. Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic double-stranded DNA virus that has been associated with a variety of human malignancies, including Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman disease. Infection with KSHV activates various DNA sensors, including cGAS, STING, IFI16, and DExD/H-box helicases. Activation of these DNA sensors induces the innate immune response to antagonize the virus. To counteract this, KSHV has developed countless strategies to evade or inhibit DNA sensing and facilitate its own infection. This review summarizes the major DNA-triggered sensing signaling pathways and details the current knowledge of DNA-sensing mechanisms involved in KSHV infection, as well as how KSHV evades antiviral signaling pathways to successfully establish latent infection and undergo lytic reactivation.


Asunto(s)
ADN Viral , Herpesvirus Humano 8 , Inmunidad Innata , Transducción de Señal , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiología , Humanos , ADN Viral/metabolismo , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/metabolismo , Sarcoma de Kaposi/virología , Nucleotidiltransferasas/metabolismo , Interacciones Huésped-Patógeno , Animales , Proteínas de la Membrana/metabolismo , Proteínas Nucleares , Fosfoproteínas
6.
J Virol ; 98(6): e0025524, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38752725

RESUMEN

Human herpesvirus 8 (HHV-8), associated with Kaposi sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman disease, encodes four interferon regulatory factor homologs, vIRFs 1-4, that interact with and inhibit various mediators of host-cell defense against virus infection. A cellular protein targeted by all the vIRFs is ubiquitin-specific protease 7 (USP7); while replication-modulatory and latently infected PEL-cell pro-viability phenotypes of USP7 targeting have been identified for vIRFs 1-3, the significance of the interaction of vIRF-4 with USP7 has remained undetermined. Here we show, through genetic ablation of the vIRF-4-USP7 interaction in infected cells, that vIRF-4 association with USP7 is necessary for optimal expression of vIRF-4 and normal HHV-8 replication. Findings from experiments on transfected and infected cells identified ubiquitination of vIRF-4 via K48-linkage and USP7-binding-associated suppression of vIRF-4 ubiquitination and, in infected cells, increased vIRF-4 expression. Analysis of IFN-I induction and associated signaling as a function of vIRF-4 and its interaction with USP7 identified a role of each in innate-immune suppression. Finally, activation via K63-polyubiquitination of the innate-immune signaling mediator TRAF3 was found to be suppressed by vIRF-4 in a USP7-binding-associated manner in infected cells, but not in transfected cells, likely via binding-regulated expression of vIRF-4. Together, our data identify the first examples of vIRF ubiquitination and a vIRF substrate of USP7, enhanced expression of vIRF-4 via its interaction with USP7, and TRAF3-inhibitory activity of vIRF-4. The findings address, for the first time, the biological significance of the interaction of vIRF-4 with USP7 and reveal a mechanism of vIRF-4-mediated innate-immune evasion and pro-replication activity via TRAF3 regulation. IMPORTANCE: HHV-8 homologs of cellular interferon regulatory factors (IRFs), involved in host-cell defense against virus infection, interact in an inhibitory fashion with IRFs and other mediators of antiviral innate immunity. These interactions are of demonstrated or hypothesized importance for successful primary, productive (lytic), and latent (persistent) infection by HHV-8. While HHV-8 vIRF-4 is known to interact physically with USP7 deubiquitinase, a key regulator of various cellular proteins, the functional and biological significance of the interaction has not been addressed. The present study identifies the interaction as important for HHV-8 productive replication and, indeed, for vIRF-4 expression and reveals a new function of vIRF-4 via inhibition of the activity of TRAF3, a pivotal mediator of host-cell antiviral activity through activation of cellular IRFs and induction of type-I interferons. These findings identify potential targets for the development of novel anti-HHV-8 agents, such as those able to disrupt vIRF-4-USP7 interaction or vIRF-4-stabilizing USP7 activity.


Asunto(s)
Herpesvirus Humano 8 , Factores Reguladores del Interferón , Peptidasa Específica de Ubiquitina 7 , Ubiquitinación , Proteínas Virales , Replicación Viral , Humanos , Herpesvirus Humano 8/fisiología , Herpesvirus Humano 8/metabolismo , Peptidasa Específica de Ubiquitina 7/metabolismo , Peptidasa Específica de Ubiquitina 7/genética , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Células HEK293 , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 3 Asociado a Receptor de TNF/genética , Unión Proteica , Interacciones Huésped-Patógeno
7.
J Virol ; 98(6): e0057624, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38767375

RESUMEN

Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus-8, is the causal agent of Kaposi sarcoma, a cancer that appears as tumors on the skin or mucosal surfaces, as well as primary effusion lymphoma and KSHV-associated multicentric Castleman disease, which are B-cell lymphoproliferative disorders. Effective prophylactic and therapeutic strategies against KSHV infection and its associated diseases are needed. To develop these strategies, it is crucial to identify and target viral glycoproteins involved in KSHV infection of host cells. Multiple KSHV glycoproteins expressed on the viral envelope are thought to play a pivotal role in viral infection, but the infection mechanisms involving these glycoproteins remain largely unknown. We investigated the role of two KSHV envelope glycoproteins, KSHV complement control protein (KCP) and K8.1, in viral infection in various cell types in vitro and in vivo. Using our newly generated anti-KCP antibodies, previously characterized anti-K8.1 antibodies, and recombinant mutant KSHV viruses lacking KCP, K8.1, or both, we demonstrated the presence of KCP and K8.1 on the surface of both virions and KSHV-infected cells. We showed that KSHV lacking KCP and/or K8.1 remained infectious in KSHV-susceptible cell lines, including epithelial, endothelial, and fibroblast, when compared to wild-type recombinant KSHV. We also provide the first evidence that KSHV lacking K8.1 or both KCP and K8.1 can infect human B cells in vivo in a humanized mouse model. Thus, these results suggest that neither KCP nor K8.1 is required for KSHV infection of various host cell types and that these glycoproteins do not determine KSHV cell tropism. IMPORTANCE: Kaposi sarcoma-associated herpesvirus (KSHV) is an oncogenic human gamma-herpesvirus associated with the endothelial malignancy Kaposi sarcoma and the lymphoproliferative disorders primary effusion lymphoma and multicentric Castleman disease. Determining how KSHV glycoproteins such as complement control protein (KCP) and K8.1 contribute to the establishment, persistence, and transmission of viral infection will be key for developing effective anti-viral vaccines and therapies to prevent and treat KSHV infection and KSHV-associated diseases. Using newly generated anti-KCP antibodies, previously characterized anti-K8.1 antibodies, and recombinant mutant KSHV viruses lacking KCP and/or K8.1, we show that KCP and K8.1 can be found on the surface of both virions and KSHV-infected cells. Furthermore, we show that KSHV lacking KCP and/or K8.1 remains infectious to diverse cell types susceptible to KSHV in vitro and to human B cells in vivo in a humanized mouse model, thus providing evidence that these viral glycoproteins are not required for KSHV infection.


Asunto(s)
Herpesvirus Humano 8 , Sarcoma de Kaposi , Proteínas del Envoltorio Viral , Proteínas Virales , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiología , Humanos , Animales , Ratones , Proteínas Virales/metabolismo , Proteínas Virales/genética , Sarcoma de Kaposi/virología , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Línea Celular , Enfermedad de Castleman/virología , Enfermedad de Castleman/metabolismo , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/metabolismo , Células HEK293 , Células Endoteliales/virología
8.
J Virol ; 98(6): e0017924, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38695538

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) belongs to the gamma-herpesvirus family and is a well-known human oncogenic virus. In infected cells, the viral genome of 165 kbp is circular DNA wrapped in chromatin. The tight control of gene expression is critical for latency, the transition into the lytic phase, and the development of viral-associated malignancies. Distal cis-regulatory elements, such as enhancers and silencers, can regulate gene expression in a position- and orientation-independent manner. Open chromatin is another characteristic feature of enhancers. To systematically search for enhancers, we cloned all the open chromatin regions in the KSHV genome downstream of the luciferase gene and tested their enhancer activity in infected and uninfected cells. A silencer was detected upstream of the latency-associated nuclear antigen promoter. Two constitutive enhancers were identified in the K12p-OriLyt-R and ORF29 Intron regions, where ORF29 Intron is a tissue-specific enhancer. The following promoters: OriLyt-L, PANp, ALTp, and the terminal repeats (TRs) acted as lytically induced enhancers. The expression of the replication and transcription activator (RTA), the master regulator of the lytic cycle, was sufficient to induce the activity of lytic enhancers in uninfected cells. We propose that the TRs that span about 24 kbp region serve as a "viral super-enhancer" that integrates the repressive effect of the latency-associated nuclear antigen (LANA) with the activating effect of RTA. Utilizing CRISPR activation and interference techniques, we determined the connections between these enhancers and their regulated genes. The silencer and enhancers described here provide an additional layer to the complex gene regulation of herpesviruses.IMPORTANCEIn this study, we performed a systematic functional assay to identify cis-regulatory elements within the genome of the oncogenic herpesvirus, Kaposi's sarcoma-associated herpesvirus (KSHV). Similar to other herpesviruses, KSHV presents both latent and lytic phases. Therefore, our assays were performed in uninfected cells, during latent infection, and under lytic conditions. We identified two constitutive enhancers, one of which seems to be a tissue-specific enhancer. In addition, four lytically induced enhancers, which are all responsive to the replication and transcription activator (RTA), were identified. Furthermore, a silencer was identified between the major latency promoter and the lytic gene locus. Utilizing CRISPR activation and interference techniques, we determined the connections between these enhancers and their regulated genes. The terminal repeats, spanning a region of about 24 kbp, seem like a "viral super-enhancer" that integrates the repressive effect of the latency-associated nuclear antigen (LANA) with the activating effect of RTA to regulate latency to lytic transition.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación Viral de la Expresión Génica , Genoma Viral , Herpesvirus Humano 8 , Regiones Promotoras Genéticas , Latencia del Virus , Herpesvirus Humano 8/genética , Humanos , Latencia del Virus/genética , Cromatina/metabolismo , Cromatina/genética , Secuencias Repetidas Terminales/genética , Replicación Viral , Células HEK293 , Antígenos Virales/genética , Antígenos Virales/metabolismo , Proteínas Nucleares
9.
J Med Virol ; 96(5): e29654, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38727099

RESUMEN

Human Herpesvirus 8 (HHV-8) has been classified by sequence analysis of open reading frame (ORF) K1, ORF K15, and variable sequence loci within the central constant region. The purpose of this study was to examine the molecular epidemiology of HHV-8 in an Irish population. This retrospective study included 30 patients who had HHV-8 DNA detected in plasma. Nested end-point PCR was used to characterise four regions of the HHV-8 genome, K1, T0.7 (K12), ORF 75, and K15. Sequencing data were obtained for 23 specimens from 19 patients. Phylogenetic analysis of ORF K1 demonstrated that subtypes A, B, C and F were present in 37%, 11%, 47% and 5%, respectively. For T0.7 and ORF 75, sequencing data were obtained for 12 patients. For T0.7, subtypes A/C, J, B, R and Q were present in 58%, 17%, 8%, 8%, and 8%, respectively. For ORF 75, subtypes A, B, C and D were present in 58%, 8%, 25%, and 8%, respectively. K15 sequences were determined for 13 patients. 69% had the P allele and 31% had the M allele. The data generated by this study demonstrate that a broad variety of HHV-8 subtypes are represented in patients exhibiting HHV-8-related disease in Ireland, a low prevalence country. The predominance of C and A K1 subtypes was as expected for a Western European population. The 31% prevalence for K15 subtype M was higher than expected for a Western European population. This may represent the changing and evolving epidemiology in Ireland due to altered migration patterns.


Asunto(s)
ADN Viral , Infecciones por Herpesviridae , Herpesvirus Humano 8 , Epidemiología Molecular , Filogenia , Análisis de Secuencia de ADN , Humanos , Irlanda/epidemiología , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/clasificación , Herpesvirus Humano 8/aislamiento & purificación , Masculino , Femenino , Estudios Retrospectivos , Persona de Mediana Edad , Adulto , ADN Viral/genética , Anciano , Adulto Joven , Reacción en Cadena de la Polimerasa , Genotipo , Adolescente , Sistemas de Lectura Abierta , Anciano de 80 o más Años , Niño , Datos de Secuencia Molecular
10.
AIDS ; 38(8): 1172-1180, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38564482

RESUMEN

OBJECTIVE: Kaposi sarcoma is a vascular tumor that affects the pulmonary system. However, the diagnosis of airway lesions suggestive of pulmonary Kaposi sarcoma (pKS) is reliant on bronchoscopic visualization. We evaluated the role of Kaposi sarcoma herpesvirus (KSHV) viral load in bronchoalveolar lavage (BAL) as a diagnostic biomarker in patients with bronchoscopic evidence of pKS and evaluated inflammatory cytokine profiles in BAL and blood samples. DESIGN: In this retrospective study, we evaluated KSHV viral load and cytokine profiles within BAL and blood samples in patients who underwent bronchoscopy for suspected pKS between 2016 and 2021. METHODS: KSHV viral load and cytokine profiles were obtained from both the circulation and BAL samples collected at the time of bronchoscopy to evaluate compartment-specific characteristics. BAL was centrifuged and stored as cell pellets and KSHV viral load was measured using primers for the KSHV K6 gene regions. RESULTS: We evaluated 38 BAL samples from 32 patients (30 with HIV co-infection) of whom 23 had pKS. In patients with airway lesions suggestive of pKS, there was higher KSHV viral load (median 3188 vs. 0 copies/10 6 cell equivalent; P  = 0.0047). A BAL KSHV viral load cutoff of 526 copies/10 6 cells had a sensitivity of 72% and specificity of 89% in determining lesions consistent with pKS. Those with pKS also had higher IL-1ß and IL-8 levels in BAL. The 3-year survival rate for pKS patients was 55%. CONCLUSION: KSHV viral load in BAL shows potential for aiding in pKS diagnosis. Patients with pKS also have evidence of cytokine dysregulation in BAL.


Asunto(s)
Líquido del Lavado Bronquioalveolar , Citocinas , Herpesvirus Humano 8 , Sarcoma de Kaposi , Carga Viral , Humanos , Sarcoma de Kaposi/virología , Sarcoma de Kaposi/diagnóstico , Herpesvirus Humano 8/aislamiento & purificación , Masculino , Femenino , Estudios Retrospectivos , Persona de Mediana Edad , Líquido del Lavado Bronquioalveolar/virología , Líquido del Lavado Bronquioalveolar/citología , Adulto , Citocinas/análisis , Broncoscopía , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/virología , Neoplasias Pulmonares/patología , Biomarcadores/análisis , Infecciones por VIH/complicaciones , Infecciones por VIH/diagnóstico , Anciano , Lavado Broncoalveolar
11.
BMC Infect Dis ; 24(1): 454, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684967

RESUMEN

BACKGROUND: Clinically, most patients with Kaposi's sarcoma (KS) are male, and several direct and indirect mechanisms may underlie this increased susceptibility in men, Kaposi's sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV-8), is considered to be the primary etiological agent responsible for KS. Thus, we propose the hypothesis that men are more susceptible to HHV-8 infection, leading to a higher incidence of Kaposi's sarcoma among males. A meta-analysis was conducted to evaluate the association between gender and HHV-8 seropositivity in the general population. METHODS: A comprehensive literature search was performed using 6 online databases: PubMed, EMBASE, Cochrane library, Web of Science, CNKI, and Wanfang. Studies published before March 15, 2023, were included. RESULTS: In all, 33 articles including 41 studies were included in the meta-analysis. In the included adult population. men had a higher risk of HHV-8 infection than did women in adult populations from all over the world (odds ratio [OR]: 1.08, 95% confidence interval [CI]: 1.01-1.15), but no differences were found in child population from all over the world (OR: 0.90, 95% CI: 0.79-1.01). There was a significant difference in HHV-8 seroprevalence between men and women in sub-Saharan Africa (SSA) adult population (OR: 1.15, 95% CI: 1.05-1.26). However, no significant differences were observed in sub-Saharan Africa (SSA) child population (OR: 0.90, 95%CI 0.78-1.03). As for other continents, the results showed no significant difference, such as the Asian population (OR: 1.03, 95%CI: 0.92-1.16). or the European and American populations (OR 1.01, 95%CI 0.87-1.17). CONCLUSION: There was a slight gender disparity for HHV-8 infection in the adult population. Among the adult populations from SSA and globally, men were more likely to be infected with HHV-8 than were women. However, no statistical significance was observed in the child populations from SSA and globally. In the future, the inclusion of more standardized studies may strengthen the results of this study.


Asunto(s)
Infecciones por Herpesviridae , Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Masculino , Femenino , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/virología , Sarcoma de Kaposi/epidemiología , Sarcoma de Kaposi/virología , Factores Sexuales , Adulto , Incidencia , Factores de Riesgo , Niño
12.
Arch Virol ; 169(5): 98, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38619650

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) causes Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman disease. The tegument is a structure that is unique to herpesviruses that includes host and viral proteins, including the viral ORF42 and ORF55 proteins. Alphaherpesvirus tegument proteins have been well studied, but much is unknown regarding KSHV. Here, we report an interaction between the ORF42 and ORF55 proteins. ORF55 interacted with and recruited ORF42 from the nucleus to the cytoplasm. When ORF42 and ORF55 were expressed simultaneously in cultured cells, the expression level of these two viral proteins was higher than when either was expressed independently. ORF55, but not ORF42, was polyubiquitinated, suggesting that an unidentified regulatory mechanism may be present. A recombinant virus with an ectopic stop codon in ORF42 exhibited normal replication of genomic DNA, but fewer virus particles were released with the recombinant than with the wild-type virus. A unique R136Q mutation in ORF42, which is found in a KSHV strain that is prevalent on Miyako Island, Okinawa Prefecture, Japan, further increased the expression of ORF42 and ORF55 when these proteins were expressed simultaneously. However, the ORF42 R136Q mutation did not affect the localization pattern of ORF42 itself or of ORF55. In addition, experiments with a recombinant virus possessing the ORF42 R136Q mutation showed lower levels of production of the mutant virus than of the wild-type virus, despite similar levels of genome replication. We suggest that the R136Q mutation in ORF42 plays an important role in ORF55 protein expression and virus production.


Asunto(s)
Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Herpesvirus Humano 8/genética , Citoplasma , Japón , Proteínas Virales/genética
14.
Cancer Res Commun ; 4(4): 1024-1040, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38592451

RESUMEN

Non-Hodgkin lymphoma (NHL) is a common cancer in both men and women and represents a significant cancer burden worldwide. Primary effusion lymphoma (PEL) is a subtype of NHL infected with Kaposi sarcoma-associated herpesvirus (KSHV). PEL is an aggressive and lethal cancer with no current standard of care, owing largely to its propensity to develop resistance to current chemotherapeutic regimens. Here, we report a reliance of KSHV-positive PEL on the mitotic kinase, NEK2, for survival. Inhibition of NEK2 with the inhibitor, JH295, resulted in caspase 3-mediated apoptotic cell death of PEL. Furthermore, NEK2 inhibition significantly prolonged survival and reduced tumor burden in a PEL mouse model. We also demonstrate that the ABC transporter proteins, MDR1 and MRP, are most active in PEL and that inhibition of NEK2 in PEL reduced the expression and activity of these ABC transporter proteins, which are known to mediate drug resistance in cancer. Finally, we report that JH295 treatment sensitized lymphomas to other chemotherapeutic agents such as rapamycin, resulting in enhanced cancer cell death. Overall, these data offer important insight into the mechanisms underlying PEL survival and drug resistance, and suggest that NEK2 is a viable therapeutic target for PEL. SIGNIFICANCE: The mitotic kinase, NEK2, is important for the survival of KSHV-positive PEL. NEK2 inhibition resulted in PEL apoptosis and reduced tumor burden in a mouse model. NEK2 inhibition also reduced drug resistance.


Asunto(s)
Herpesvirus Humano 8 , Linfoma no Hodgkin , Linfoma de Efusión Primaria , Masculino , Animales , Ratones , Humanos , Femenino , Linfoma de Efusión Primaria/tratamiento farmacológico , Transportadoras de Casetes de Unión a ATP , Agresión , Modelos Animales de Enfermedad , Quinasas Relacionadas con NIMA/genética
15.
Int J Mol Sci ; 25(7)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38612584

RESUMEN

Human herpesvirus 8 (HHV8)-associated diseases include Kaposi sarcoma (KS), multicentric Castleman disease (MCD), germinotropic lymphoproliferative disorder (GLPD), Kaposi sarcoma inflammatory cytokine syndrome (KICS), HHV8-positive diffuse large B-cell lymphoma (HHV8+ DLBCL), primary effusion lymphoma (PEL), and extra-cavitary PEL (ECPEL). We report the case of a human immunodeficiency virus (HIV)-negative male treated for cutaneous KS, who developed generalized lymphadenopathy, hepatosplenomegaly, pleural and abdominal effusions, renal insufficiency, and pancytopenia. The excised lymph node showed features of concomitant involvement by micro-KS and MCD, with aggregates of HHV8+, Epstein Barr virus (EBV)-negative, IgM+, and lambda+ plasmablasts reminiscent of microlymphoma. Molecular investigations revealed a somatically hypermutated (SHM) monoclonal rearrangement of the immunoglobulin heavy chain (IGH), accounting for 4% of the B-cell population of the lymph node. Mutational analyses identified a pathogenic variant of KMT2D and variants of unknown significance in KMT2D, FOXO1, ARID1A, and KMT2A. The patient died shortly after surgery. The histological features (HHV8+, EBV-, IgM+, Lambda+, MCD+), integrated with the molecular findings (monoclonal IGH, SHM+, KMT2D mutated), supported the diagnosis of a monoclonal HHV8+ microlymphoma, with features intermediate between an incipient HHV8+ DLBCL and an EBV-negative ECPEL highlighting the challenges in the accurate classification of HHV8-driven lymphoid proliferations.


Asunto(s)
Enfermedad de Castleman , Infecciones por Virus de Epstein-Barr , Infecciones por VIH , Herpesvirus Humano 8 , Sarcoma de Kaposi , Masculino , Humanos , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/genética , Herpesvirus Humano 4 , Infecciones por VIH/complicaciones , Inmunoglobulina M
16.
PLoS Pathog ; 20(4): e1012141, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38626263

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is a double-stranded DNA virus etiologically associated with multiple malignancies. Both latency and sporadic lytic reactivation contribute to KSHV-associated malignancies, however, the specific roles of many KSHV lytic gene products in KSHV replication remain elusive. In this study, we report that ablation of ORF55, a late gene encoding a tegument protein, does not impact KSHV lytic reactivation but significantly reduces the production of progeny virions. We found that cysteine 10 and 11 (C10 and C11) of pORF55 are palmitoylated, and the palmytoilation is essential for its Golgi localization and secondary envelope formation. Palmitoylation-defective pORF55 mutants are unstable and undergo proteasomal degradation. Notably, introduction of a putative Golgi localization sequence to these palmitoylation-defective pORF55 mutants restores Golgi localization and fully reinstates KSHV progeny virion production. Together, our study provides new insight into the critical role of pORF55 palmitoylation in KSHV progeny virion production and offers potential therapeutic targets for the treatment of related malignancies.


Asunto(s)
Aparato de Golgi , Herpesvirus Humano 8 , Lipoilación , Proteínas Virales , Virión , Replicación Viral , Herpesvirus Humano 8/fisiología , Herpesvirus Humano 8/metabolismo , Aparato de Golgi/metabolismo , Aparato de Golgi/virología , Humanos , Virión/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/genética , Replicación Viral/fisiología , Células HEK293
17.
Cell Host Microbe ; 32(5): 755-767.e4, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38653242

RESUMEN

Kaposi sarcoma (KS) is the most common cancer in persons living with HIV. It is caused by KS-associated herpesvirus (KSHV). There exists no animal model for KS. Pronuclear injection of the 170,000-bp viral genome induces early-onset, aggressive angiosarcoma in transgenic mice. The tumors are histopathologically indistinguishable from human KS. As in human KS, all tumor cells express the viral latency-associated nuclear antigen (LANA). The tumors transcribe most viral genes, whereas endothelial cells in other organs only transcribe the viral latent genes. The tumor cells are of endothelial lineage and exhibit the same molecular pattern of pathway activation as KS, namely phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR, interleukin-10 (IL-10), and vascular endothelial growth factor (VEGF). The KSHV-induced tumors are more aggressive than Ha-ras-induced angiosarcomas. Overall survival is increased by prophylactic ganciclovir. Thus, whole-virus KSHV-transgenic mice represent an accurate model for KS and open the door for the genetic dissection of KS pathogenesis and evaluation of therapies, including vaccines.


Asunto(s)
Modelos Animales de Enfermedad , Hemangiosarcoma , Herpesvirus Humano 8 , Ratones Transgénicos , Sarcoma de Kaposi , Animales , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidad , Ratones , Hemangiosarcoma/virología , Hemangiosarcoma/genética , Hemangiosarcoma/patología , Sarcoma de Kaposi/virología , Sarcoma de Kaposi/patología , Genoma Viral , Humanos , Antígenos Virales/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Ganciclovir/uso terapéutico , Ganciclovir/farmacología , Interleucina-10/genética
18.
J Cancer Res Ther ; 20(1): 469-471, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38554367

RESUMEN

Kaposi's sarcoma (KS) is an angiogenic tumor. KS lesions frequently develop in the skin and oral cavity mucosa in the head and neck regions, and pure laryngeal localization is extremely rare. We reported a 64-year-old male patient without HIV, HBV, and HCV positivity presented with a hemangiomatous lesion detected incidentally in the right vocal cord. Biopsy was taken for histopathological and immunohistochemical evaluation. Examination revealed that spindle cells were of vascular origin and expressed HHV-8, a specific marker associated with Kaposi's sarcoma-associated herpesvirus. Positron emission tomography-computed tomography (PET/CT) demonstrated an increased fluorodeoxyglucose (FDG) uptake in the vocal cord. The patient was treated with a 30 Gy volumetric arc plan. Disease-free follow-up continues in the first year after low-dose definitive RT. This is the first case report of KS in the vocal cord in which the most detailed data about RT were shared.


Asunto(s)
Herpesvirus Humano 8 , Sarcoma de Kaposi , Masculino , Humanos , Persona de Mediana Edad , Sarcoma de Kaposi/diagnóstico , Sarcoma de Kaposi/radioterapia , Pliegues Vocales , Tomografía Computarizada por Tomografía de Emisión de Positrones , Piel/patología
19.
Rinsho Ketsueki ; 65(2): 74-77, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-38448001

RESUMEN

An 80-year-old Japanese man presented with systemic lymphadenopathy, including the para-aortic area and left inguinal nodes, which was diagnosed as diffuse large B-cell lymphoma (DLBCL) and human herpesvirus (HHV) 8-positive/HIV-negative Kaposi's sarcoma (KS). Immunohistochemical examination revealed that the lymphoma cells were negative for HHV-8. The patient received combined chemotherapy with rituximab, pirarubicin, cyclophosphamide, vincristine, and prednisolone for six cycles and achieved complete remission. In the literature, five cases of simultaneous appearance of malignant lymphoma and KS in the same lymph node have been reported, but DLBCL as a histological subtype has not yet been reported.


Asunto(s)
Herpesvirus Humano 8 , Linfoma de Células B Grandes Difuso , Sarcoma de Kaposi , Masculino , Humanos , Anciano de 80 o más Años , Sarcoma de Kaposi/complicaciones , Sarcoma de Kaposi/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/complicaciones , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Ganglios Linfáticos , VIH
20.
Front Immunol ; 15: 1361009, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38482011

RESUMEN

The development of lymphoma is a complex multistep process that integrates numerous experimental findings and clinical data that have not yet yielded a definitive explanation. Studies of oncogenic viruses can help to deepen insight into the pathogenesis of lymphoma, and identifying associations between lymphoma and viruses that are established and unidentified should lead to cellular and pharmacologically targeted antiviral strategies for treating malignant lymphoma. This review focuses on the pathogenesis of lymphomas associated with hepatitis B and C, Epstein-Barr, and human immunodeficiency viruses as well as Kaposi sarcoma-associated herpesvirus to clarify the current status of basic information and recent advances in the development of virus-associated lymphomas.


Asunto(s)
Herpesvirus Humano 8 , Linfoma , Humanos , Linfoma/etiología , Virus Oncogénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA